A Dipolar Cycloaddition Reaction To Access 6-Methyl-4,5,6,7

Dec 6, 2017 - A single pot dipolar cycloaddition reaction/Cope elimination ... to the initiation of proof of concept studies for the treatment of mood...
0 downloads 0 Views 3MB Size
Article pubs.acs.org/jmc

Cite This: J. Med. Chem. 2018, 61, 207−223

A Dipolar Cycloaddition Reaction To Access 6‑Methyl-4,5,6,7tetrahydro‑1H‑[1,2,3]triazolo[4,5‑c]pyridines Enables the Discovery Synthesis and Preclinical Profiling of a P2X7 Antagonist Clinical Candidate Christa C. Chrovian,*,† Akinola Soyode-Johnson,† Alexander A. Peterson,† Christine F. Gelin,† Xiaohu Deng,† Curt A. Dvorak,† Nicholas I. Carruthers,† Brian Lord,† Ian Fraser,† Leah Aluisio,† Kevin J. Coe,† Brian Scott,† Tatiana Koudriakova,† Freddy Schoetens,† Kia Sepassi,† David J. Gallacher,‡ Anindya Bhattacharya,† and Michael A. Letavic† †

Janssen Research & Development, LLC, 3210 Merryfield Row, San Diego, California 92121, United States Janssen Research & Development, Janssen Pharmaceutica NV, Turnhoutseweg 30, 2340 Beerse, Belgium



S Supporting Information *

ABSTRACT: A single pot dipolar cycloaddition reaction/Cope elimination sequence was developed to access novel 1,4,6,7tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridine P2X7 antagonists that contain a synthetically challenging chiral center. The structure−activity relationships of the new compounds are described. Two of these compounds, (S)-(2-fluoro-3-(trifluoromethyl)phenyl)(1-(5-fluoropyrimidin-2-yl)-6-methyl-1,4,6,7tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridin-5-yl)methanone (compound 29) and (S)-(3-fluoro-2-(trifluoromethyl)pyridin-4yl)(1-(5-fluoropyrimidin-2-yl)-6-methyl-1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridin-5-yl)methanone (compound 35), were found to have robust P2X7 receptor occupancy at low doses in rat with ED50 values of 0.06 and 0.07 mg/kg, respectively. Compound 35 had notable solubility compared to 29 and showed good tolerability in preclinical species. Compound 35 was chosen as a clinical candidate for advancement into phase I clinical trials to assess safety and tolerability in healthy human subjects prior to the initiation of proof of concept studies for the treatment of mood disorders.



INTRODUCTION Proinflammatory cytokines may be a significant contributor to the pathology of psychiatric malfunctioning. Indeed, elevated levels of inflammatory biomarkers have been identified in individuals diagnosed with bipolar and major depressive disorders.1 The P2X7 receptor is an ATP-gated ion channel involved in a prominent inflammatory cascade in central nervous system (CNS) immune cells (microglia) that results in the release of the “master” inflammatory cytokine IL-1β, so-called because of its role in triggering an acute phase response.2 P2X7 enables the release of IL-1β by converting pro-IL-1β into its mature form. In the CNS this occurs primarily in microglia and triggers a neuroinflammatory cascade. As part of our ongoing interest in the chronic effects of prolonged neuroinflammation, and in particular the role of neuroinflammation in psychiatric conditions, we have been working to advance brain-penetrant selective P2X7 antagonists into clinical proof of concept (PoC) studies in order to study the effect of inhibiting overall IL-1β release (CNS + peripheral) on individuals suffering from mood disorders. Toward this goal, we have found that P2X7 antagonists are efficacious in preclinical chronic models of depression.3 Data have also been presented suggesting that P2X7 antagonists that can cross the blood−brain barrier may be the most effective at treating psychiatric conditions.4,5 © 2017 American Chemical Society

At least two selective P2X7 antagonists have advanced into PoC studies for the treatment of Crohn’s disease (CD)6 and/or rheumatoid arthritis (RA).7,8 These two compounds (one from Pfizer8 and one from AstraZeneca6,7) were both effective at blocking peripheral IL-1β release in ex vivo ATP-activated patient plasma samples. Neither compound ultimately met its desired clinical end point in the studies, and their development for either RA7,8 and/or CD6 was discontinued. We recently disclosed the structure of JNJ-54175446 (1, Chart 1), which has successfully completed initial phase I clinical studies.9 Compound 1 is a potent P2X7 antagonist with significant brain penetration and exhibits dose-dependent P2X7 receptor occupancy in the hippocampus of rats and dogs. It is considered to be a Biopharmaceutical Classification System (BCS) class II compound based on its high permeability and low solubility; in aqueous buffer its solubility was 39 μM at pH 2 and 21 μM at pH 7 but was found to be readily absorbed in preclinical species, with a bioavailability of ∼100% in dog. The predicted human half-life of 1 was 29 h.9 While compound 1 progressed into preclinical development, the project team remained interested in a backup compound with Received: August 28, 2017 Published: December 6, 2017 207

DOI: 10.1021/acs.jmedchem.7b01279 J. Med. Chem. 2018, 61, 207−223

Journal of Medicinal Chemistry



Chart 1. Fused Triazole and Pyrimidine Core P2X7 Antagonists Containing a C-Methyl or -Phenyl Substituent in the Piperidine or Piperazine Ring

Article

RESULTS AND DISCUSSION

Synthetic Methodology Development. One challenge in executing this proposal was devising synthetic methods for the rapid assembly of final compounds. Compelling 1,2,3-triazole syntheses includes click chemistry of an alkyne and an azide.16 In our case of a fused triazole, this would be followed by elaborating the 5-position of the ring with functionality that could, after a ring closing reaction with a 4-methylamino group, form the fused triazole system (Scheme 1A).17 However, this strategy is rarely used with Het- or aryl-azides and becomes more challenging given the 6-methyl group we needed to install.17−19 In an alternative method, the fused triazole can be formed from a diaminopyridine, which is followed by a reduction of the pyridine ring (Scheme 1B).13,20 Both of these routes require several challenging steps. With the required absolute configuration, the synthesis becomes additionally arduous. We were interested in developing a concise route to enable rapid access to the 1-Het-6S-methyl1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c] pyridines and thought that a 1,3-dipolar cycloaddition of an azide and a ketone was worth investigating. As early as 1967, Huisgen et al. reported 1,3-dipolar cycloadditions with azides and an enamide that could form triazoles in a fused ring system.21 This reaction has been more recently elaborated using ketones and azides in the presence of an amine organocatalyst. In one example, Ramachary and co-workers reported the use of activated ketones in an enamine amination/ elimination followed by [3 + 2]-cycloaddition/hydrolysis in an organocatalytic cascade as a synthetic approach to substituted 1,2,3-triazoles (Scheme 2A).22 Perhaps more relevant to our work is the cascade reaction using unactivated ketones reported in 2011 by Belkheira et al.23 In this work, carbocyclic ketones were reacted with aryl azides, presumably via enamine formation, in a dipolar cycloaddition reaction (Scheme 2B). We hoped we could extend this methodology to the formation of 6-methyl4,5,6,7-tetrahydro-1H-[1,2,3]triazolo[4,5-c]pyridines from unsymmetrical ketone heterocycles containing a chiral center (Scheme 2C). We kept in mind, however, that the altered electronics and added steric bulk of our starting ketone (versus the literature), as well as the need for a specific regiochemical outcome, could make the optimization challenging. Initial investigations were conducted using phenyl azide and ketone rac-6 following the protocol reported by Belkheira et al.23 We found that the reaction did not proceed with 6 using their optimized conditions of catalytic proline and CH2Cl2 solvent (Table 1, entry 1). We found that when pyrrolidine was used instead of proline, the reaction proceeded, albeit in low yields (entry 2). However, the products that were isolated contained a

high solubility as well as differentiated absorption, distribution, metabolism, and elimination (ADME) properties, specifically, one with more rapid elimination. Throughout our work on P2X7 antagonists containing 5,6-fused cores,10 we have found that a 4R-methyl (as in 19 and 211) or -phenyl (as in 3)12 substituent provides improved potency compared to an unsubstituted 4-position,10,13 particularly in rodent species. The added 4R-substituent also provided improved microsomal stability9,11 and decreased CYP2C19 inhibition.9 The recent report describing compound 1 establishes that a methyl 4R-orientation is the preferred epimer for P2X7 potency of 4-methyl-4,5,6,7-tetrahydro-1H-[1,2,3]triazolo[4,5-c]pyridines.9 Separately, we have shown that 6-methyl groups also provide benefit in fused ring cores (414 and 515), and the stereochemistry was determined to be S in the examples for which an assignment was conclusively made, including 414 and 1-(5,6-dihydro[1,2,4]triazolo[4,3-a]pyrazin-7(8H)-yl)methanones.10 It should be noted that while the absolute configuration of 5 was not determined, the specific rotation value was positive.15 The 6S-methyl group had been underexplored with 4,5,6,7-tetrahydro-1H-[1,2,3]triazolo[4,5-c]pyridine cores. We thus became interested in investigating (S)-6-methyl-1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridines.

Scheme 1. Synthetic Methods for the Preparation of 1-Het-1,4,6,7-tetrahydro-5H-[1,2,3]triazolo[4,5-c]pyridines

208

DOI: 10.1021/acs.jmedchem.7b01279 J. Med. Chem. 2018, 61, 207−223

Journal of Medicinal Chemistry

Article

after formation of the intermediates 7 and 8 in roughly 2 h, m-CPBA was added. Due to this development, our experimental procedure for forming fused triazoles was now quite simple in that three steps, (i) enamine formation, (ii) dipolar cycloaddition, and (iii) Cope elimination, are conducted in a single pot. However, an unresolved problem remained for the formation of phenyl-substituted 6-methyl-4,5,6,7-tetrahydro-1H-[1,2,3]triazolo[4,5-c]pyridines. There was no regioselectivity for the formation of 7a versus 8a. Since the reaction appeared to have many variables that could potentially be optimized, we undertook a more detailed investigation of nitrogen protecting groups, amine bases, solvents, and temperatures. The next reaction optimizations were conducted using 2-azido-5-fluoropyrimidine (the same pyrimidine featured in 1) and N-protected 2-methylpiperidones (Table 2). A solvent and temperature screen revealed that the best yields and regioselectivities were obtained at higher temperatures with toluene or 1,4-dioxane (entries 1−6). Dean−Stark conditions had minimal impact on the regioselectivity and yield (entry 7). It is interesting to note that using phenyl azide under the best reaction conditions resulted in a significantly lower regioselectivity than when the fluoropyrimidine azide was used (compare entry 1, Table 2 (88:12) with entry 5, Table 1 (1:1)). This is a curious and unexpected finding. One possible reason for this could be that the electron deficiency of the fluoropyrimidine azide favors a thermodynamic product by enabling reversibility, although this hypothesis has not been tested. The nitrogen protecting groups were also screened, and carbamate protecting groups worked well but N-methyl and -benzyl groups resulted in no reaction (entries 8−11). A screen of secondary amines appeared to favor the use of unsubstituted pyrrolidine (a subset of amines is shown in entries 12−16). Proline once again did not promote the reaction (entry 14). The scope of the reaction was investigated using a variety of carbocyclic and heterocyclic ketones, as well as aryl and alkyl azides. The reaction’s utility is enhanced by the fact that both

Scheme 2. Synthesis of Fused 1,2,3-Triazoles by an Enamine Amination/Elimination Followed by [3 + 2]-Cycloaddition/ Hydrolysis Organocatalytic Cascade

pyrrolidine adduct (7a and 8a), which we supposed was an intermediate that could be readily converted to the desired product. It was not surprising that the yield improved when a full equivalent of pyrrolidine was used (entry 3). Comparable yields were achieved with ethanol solvent (entry 4) and were higher using toluene (entry 5). Unfortunately, attempts to facilitate an acid catalyzed elimination of 7a + 8a to form 9a + 10a were unsuccessful using either catalytic or stoichiometric amounts of p-TSA. One thought we had was to use oxidative conditions in order to set up a Cope elimination. Gratifyingly, after treating an isolated mixture of 7a + 8a (1:1) with m-CPBA, a rapid transformation occurred to form 9a + 10a in a 1:1 ratio and 83% isolated yield (Scheme 3). In fact, this elimination could even be conducted in a single pot format, following the [3 + 2] cycloaddition step. The best overall yields for the sequence were obtained when we combined the ketone, azide, and pyrrolidine at once with heating. Then,

Table 1. Initial Results of a Dipolar Cycloaddition with 2-Methylpiperidone and Phenyl Azide

a

entry

amine

solvent

conditions

isolated yielda

1 2 3 4 5

proline, catalytic pyrrolidine, catalytic pyrrolidine, 1 equiv pyrrolidine, 1 equiv pyrrolidine, 1 equiv

DCM DCM DCM EtOH toluene

rt or 80 °C, microwave 80 °C, microwave 80 °C, microwave 120 °C, microwave 100 °C

no reaction